Page 161 - 2019_09-HaematologicaMondo-web
P. 161

Characterizing energy metabolism of CLL
in line with our observation.41
In our study, we also highlighted the possibility of
exploiting heterogeneity of energy metabolism to improve individualized patient care. We show that higher glycolytic flexibility can contribute to the resistance of CLL samples to treatment with drugs that affect mito- chondria, such as rotenone, venetoclax, and navitoclax. We postulate that the cytotoxic effects of these drugs may partially result from restricting the energy supply by blocking cellular respiration and thus, cells with higher glycolytic potential can counteract their effect due to high- er metabolic flexibility.
The current study has certain limitations. Firstly, while most of the proliferative activity of CLL cells appears in lymph node and bone marrow, in this study we only used circulating CLL cells due to the easier availability of patient material, which was instrumental in providing an adequate study size. In addition, although we observed many biologically meaningful associations, these are gen- erally weak, as indicated by the relatively small effect sizes or correlation coefficients. While it is possible that biological variables not measured by us contribute to the heterogeneity in energy metabolism, a likely explanation could be biological noise (since we are using patient sam- ples instead of cell lines) and technical noise of the Seahorse extracellular flux measurements, and the other
assays used. Indeed, our study is, to our knowledge, the first that uses such a dynamic assay to systematically interrogate energy metabolism on such a large scale.
Taken together, our in-depth characterization of energy metabolism and integrative analyses provide valuable insights on mechanisms underlying the metabolic regula- tion of CLL cells, and reveal the possibilities of guiding clinical diagnosis and individualized patient care based on metabolic profiles. Our large-scale energy metabolism dataset complements the current traditional omics datasets, such as RNA sequencing, DNA sequencing, and methylation profiling, and contribute to a better under- standing of CLL biology.
Acknowledgments
The authors thank the reviewers for helpful suggestions and comments, which improved the quality of this work.
Funding
The work was supported by the European Union (Horizon 2020 project SOUND) and GCH-CLL project co-founded by the European Commission/DG Research and Innovation. DM was supported by the Else Kröner-Fresenius-Stiftung. DM and MB were supported by the Erich und Gertrud Roggenbuck- Stiftung. TZ was supported by the Monique Dornonville de la Cour Stiftung and the Cancer Research Center (CRC) Zurich.
References
1. MessmerBT,MessmerD,AllenSL,etal.In vivo measurements document the dynamic cellular kinetics of chronic lymphocytic leukemia B cells. J Clin Invest. 2005; 115(3):755-764.
2. vanGentR,KaterAP,OttoSA,etal.Invivo Dynamics of Stable Chronic Lymphocytic Leukemia Inversely Correlate with Somatic Hypermutation Levels and Suggest No Major Leukemic Turnover in Bone Marrow. Cancer Res. 2008;68(24):10137-10144.
3. Hanahan D, Weinberg RA. Hallmarks of cancer: the next generation. Cell. 2011; 144(5):646-674.
4. Garcia-Manteiga JM, Mari S, Godejohann M, et al. Metabolomics of B to plasma cell differentiation. J Proteome Res. 2011; 10(9):4165-4176.
5. MoranEC,KamigutiAS,CawleyJC,Pettitt AR. Cytoprotective antioxidant activity of serum albumin and autocrine catalase in chronic lymphocytic leukaemia. Br J Haematol. 2002;116(2):316-328.
6. Jitschin R, Hofmann AD, Bruns H, et al. Mitochondrial metabolism contributes to oxidative stress and reveals therapeutic tar- gets in chronic lymphocytic leukemia. Blood. 2014;123(17):2663v2672.
7. MacIntyre DA, Jiménez B, Lewintre EJ, et al. Serum metabolome analysis by 1H- NMR reveals differences between chronic lymphocytic leukaemia molecular sub- groups. Leukemia. 2010;24(4):788-797.
8. Zenz T, Mertens D, Küppers R, Döhner H, Stilgenbauer S. From pathogenesis to treat- ment of chronic lymphocytic leukaemia. Nat Rev Cancer. 2010;10(1):37-50.
9. Fabbri G, Dalla-Favera R. The molecular pathogenesis of chronic lymphocytic leukaemia. Nat Rev Cancer. 2016;1
6(3):145-162.
10. Dietrich S, Oleś M, Lu J, et al. Drug-pertur-
bation-based stratification of blood cancer.
J Clin Invest. 2018;128(1):427-445.
11. Wu S-H, Bi J-F, Cloughesy T, Cavenee WK, Mischel PS. Emerging function of mTORC2 as a core regulator in glioblastoma: metabol- ic reprogramming and drug resistance.
Cancer Biol Med. 2014; 11(4):255-263.
12. Long Y, Tsai W-B, Wangpaichitr M, et al. Arginine Deiminase Resistance in Melanoma Cells Is Associated with Metabolic Reprogramming, Glucose Dependence, and Glutamine Addiction.
Mol Cancer Ther. 2013;12(11):2581-2590.
13. Böttcher M, Renner K, Berger R, et al. D-2- hydroxyglutarate interferes with HIF-1a stability skewing T-cell metabolism towards oxidative phosphorylation and impairing Th17 polarization.
Oncoimmunology. 2018;e1445454.
14. Oakes CC, Seifert M, Assenov Y, et al. DNA methylation dynamics during B cell maturation underlie a continuum of dis- ease phenotypes in chronic lymphocytic
leukemia. Nat Genet. 2016;48(3):253-264.
15. Liberzon A, Birger C, Thorvaldsdóttir H, Ghandi M, Mesirov JP, Tamayo P. The Molecular Signatures Database (MSigDB) hallmark gene set collection. Cell Syst.
2015;1(6):417-425.
16. Semenza GL, Roth PH, Fang HM, Wang
GL. Transcriptional regulation of genes encoding glycolytic enzymes by hypoxia- inducible factor 1. J Biol Chem. 1994; 269(38):23757-23763.
17. Hitosugi T, Zhou L, Elf S, et al. Phosphoglycerate Mutase 1 Coordinates Glycolysis and Biosynthesis to Promote Tumor Growth. Cancer Cell. 2012; 22(5):585-600.
18. Dunaway GA, Kasten TP, Sebo T, Trapp R.
Analysis of the phosphofructokinase sub- units and isoenzymes in human tissues. Biochem J. 1988;251(3):677-683.
19. Stevenson FK, Krysov S, Davies AJ, Steele AJ, Packham G. B-cell receptor signaling in chronic lymphocytic leukemia. Blood. 2011;118(16):4313-4320.
20. Tavolaro S, Colombo T, Chiaretti S, et al. Increased chronic lymphocytic leukemia proliferation upon IgM stimulation is sus- tained by the upregulation of miR-132 and miR-212. Genes Chromosom Cancer. 2015; 54(4):222-234.
21. Doughty CA, Bleiman BF, Wagner DJ, et al. Antigen receptor-mediated changes in glu- cose metabolism in B lymphocytes: Role of phosphatidylinositol 3-kinase signaling in the glycolytic control of growth. Blood. 2006;107(11):4458-4465.
22. Qorraj M, Bruns H, Böttcher M, et al. The PD-1/PD-L1 axis contributes to immune metabolic dysfunctions of monocytes in chronic lymphocytic leukemia. Leukemia. 2017;31(2):470-478.
23. Lagadinou ED, Sach A, Callahan K, et al. BCL-2 inhibition targets oxidative phos- phorylation and selectively eradicates qui- escent human leukemia stem cells. Cell Stem Cell. 2013;12(3):329-341.
24. Pallasch CP, Schwamb J, Königs S, et al. Targeting lipid metabolism by the lipopro- tein lipase inhibitor orlistat results in apop- tosis of B-cell chronic lymphocytic leukemia cells. Leukemia. 2008;22(3):585-592.
25. Ahn IE, Underbayev C, Albitar A, et al. Clonal evolution leading to ibrutinib resist- ance in chronic lymphocytic leukemia. Blood. 2017;129(11):1469-1479.
26. Udensi UK, Tchounwou PB. Dual effect of oxidative stress on leukemia cancer induc- tion and treatment. J Exp Clin Cancer Res. 2014;33:106.
haematologica | 2019; 104(9)
1839


































































































   159   160   161   162   163